Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Int J Cancer ; 134(10): 2330-41, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24154973

ABSTRACT

A locus on human chromosome 11q23 tagged by marker rs3802842 was associated with colorectal cancer (CRC) in a genome-wide association study; this finding has been replicated in case-control studies worldwide. In order to identify biologic factors at this locus that are related to the etiopathology of CRC, we used microarray-based target selection methods, coupled to next-generation sequencing, to study 103 kb at the 11q23 locus. We genotyped 369 putative variants from 1,030 patients with CRC (cases) and 1,061 individuals without CRC (controls) from the Ontario Familial Colorectal Cancer Registry. Two previously uncharacterized genes, COLCA1 and COLCA2, were found to be co-regulated genes that are transcribed from opposite strands. Expression levels of COLCA1 and COLCA2 transcripts correlate with rs3802842 genotypes. In colon tissues, COLCA1 co-localizes with crystalloid granules of eosinophils and granular organelles of mast cells, neutrophils, macrophages, dendritic cells and differentiated myeloid-derived cell lines. COLCA2 is present in the cytoplasm of normal epithelial, immune and other cell lineages, as well as tumor cells. Tissue microarray analysis demonstrates the association of rs3802842 with lymphocyte density in the lamina propria (p = 0.014) and levels of COLCA1 in the lamina propria (p = 0.00016) and COLCA2 (tumor cells, p = 0.0041 and lamina propria, p = 6 × 10(-5)). In conclusion, genetic, expression and immunohistochemical data implicate COLCA1 and COLCA2 in the pathogenesis of colon cancer. Histologic analyses indicate the involvement of immune pathways.


Subject(s)
Colon/metabolism , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease/genetics , Immune System/metabolism , Polymorphism, Single Nucleotide , Amino Acid Sequence , Blotting, Western , Caco-2 Cells , Cell Line, Tumor , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Genetic Predisposition to Disease/classification , HCT116 Cells , HEK293 Cells , HL-60 Cells , HT29 Cells , HeLa Cells , Humans , Immune System/pathology , Immunohistochemistry , Jurkat Cells , K562 Cells , MCF-7 Cells , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phylogeny , RNA , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , U937 Cells
2.
Pharm Res ; 28(12): 3079-90, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21818714

ABSTRACT

PURPOSE: MicroRNA-101 (miR-101) expression is negatively associated with tumor growth and proliferation in several solid epithelial cancers. Enhancer of zeste homolog 2 (EzH2) appears to be a functional target of miR-101. We explore the role of miR-101 and its interaction with EzH2 in epithelial ovarian carcinoma (EOC). METHODS: In situ hybridization (ISH) for miR-101 was performed on EOC patient tissues and normal controls. EOC cell lines were transfected with miR-101 and subjected to growth analysis and clonogenic assays. Cell motility was assessed by Boyden chamber and wound-healing assays. P21(waf1/cip1) and EzH2 interaction was assessed by Chromatin Immunoprecipitation (ChIP) assay in MDAH-2774 cells. SCID mice were assessed for tumor burden after injection with miR-101 or control vector-treated MDAH-2774 cells. RESULTS: ISH analysis revealed a decrease in miR-101 expression in EOC compared with normal tissue. MiR-101 re-expression in EOC cell lines resulted in increased apoptosis, decreased cellular proliferation, invasiveness, and reduced growth of tumor xenografts. CHIP assays revealed that re-expression of miR-101 inhibited the interaction of EzH2 with p21(waf1/cip1) promoter. CONCLUSIONS: MiR-101 re-expression appears to have antitumor effects, providing a better understanding of the role of miR-101 in EOC.


Subject(s)
Chromatin/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/genetics , MicroRNAs/genetics , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Transcription Factors/genetics , Animals , Apoptosis , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, SCID , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Ovary/metabolism , Ovary/pathology , Polycomb Repressive Complex 2 , Transcription Factors/metabolism
3.
Curr Pharm Biotechnol ; 22(8): 1122-1128, 2021.
Article in English | MEDLINE | ID: mdl-33023442

ABSTRACT

BACKGROUND: Aloe vera has been reported as a topical antibiotic and healing agent for wounds, but advantages of oral administration and mechanisms of wound healing have not been reported. Present study focuses on the evaluation of effects of oral administration of Aloe vera for excisional cutaneous wounds in Sprague Dawley rats. METHODS: Sprague Dawley (SD) rats were inflicted with excisional wounds and were either treated with Aloe vera orally (Aloe vera) or kept untreated (wound). In contrast, healthy rats were kept as control group. Wound area was measured from day 7th to day 21st. Collagen content was estimated by hydroxyproline assay. Histology was analysed by hematoxylin and eosin staining. Angiogenesis was observed by indirect ELISA for Insulin like Growth Factor (IGF-1) and Vascular Endothelial Growth Factor (VEGF) protein from skin, serum and bone marrow. Chemotaxis was evaluated by RT-PCR analysis for Stromal cell-Derived Factor-1 (SDF-1) and C-X-C chemokine receptor type 4 (CXCR-4) from skin and bone marrow. RESULTS: Aloe vera healed wounds earlier than untreated rats with gradual improvement in wound areas and collagen content. Aloe vera also improved the expression of IGF-1 and VEGF in skin and bone marrow indicating an improvement in angiogenesis. RT- PCR analysis showed increased expression of genes for chemotaxis (SDF-1 and CXCR-4) in skin and bone marrow. CONCLUSION: Aloe vera improves healing by increasing collagen content, improving angiogenesis and chemotaxis.


Subject(s)
Aloe , Neovascularization, Physiologic/drug effects , Plant Extracts/pharmacology , Wound Healing/drug effects , Administration, Oral , Animals , Chemokine CXCL12/drug effects , Chemotaxis/drug effects , Collagen/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, CXCR4/drug effects , Skin/drug effects , Somatomedins/drug effects , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism
4.
Mol Cancer ; 9: 47, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20196847

ABSTRACT

BACKGROUND: Sulforaphane (SFN), an isothiocyanate phytochemical present predominantly in cruciferous vegetables such as brussels sprout and broccoli, is considered a promising chemo-preventive agent against cancer. In-vitro exposure to SFN appears to result in the induction of apoptosis and cell-cycle arrest in a variety of tumor types. However, the molecular mechanisms leading to the inhibition of cell cycle progression by SFN are poorly understood in epithelial ovarian cancer cells (EOC). The aim of this study is to understand the signaling mechanisms through which SFN influences the cell growth and proliferation in EOC. RESULTS: SFN at concentrations of 5-20 microM induced a dose-dependent suppression of growth in cell lines MDAH 2774 and SkOV-3 with an IC50 of ~8 microM after a 3 day exposure. Combination treatment with chemotherapeutic agent, paclitaxel, resulted in additive growth suppression. SFN at ~8 microM decreased growth by 40% and 20% on day 1 in MDAH 2774 and SkOV-3, respectively. Cells treated with cytotoxic concentrations of SFN have reduced cell migration and increased apoptotic cell death via an increase in Bak/Bcl-2 ratio and cleavage of procaspase-9 and poly (ADP-ribose)-polymerase (PARP). Gene expression profile analysis of cell cycle regulated proteins demonstrated increased levels of tumor suppressor retinoblastoma protein (RB) and decreased levels of E2F-1 transcription factor. SFN treatment resulted in G1 cell cycle arrest through down modulation of RB phosphorylation and by protecting the RB-E2F-1 complex. CONCLUSIONS: SFN induces growth arrest and apoptosis in EOC cells. Inhibition of retinoblastoma (RB) phosphorylation and reduction in levels of free E2F-1 appear to play an important role in EOC growth arrest.


Subject(s)
Cell Cycle/drug effects , E2F1 Transcription Factor/metabolism , Epithelial Cells/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Retinoblastoma Protein/metabolism , Thiocyanates/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA, Neoplasm/biosynthesis , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isothiocyanates , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , Paclitaxel/pharmacology , Phosphorylation/drug effects , S Phase/drug effects , Sulfoxides , Wound Healing/drug effects
5.
Mol Endocrinol ; 20(8): 1912-23, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16556730

ABSTRACT

Prolactin (PRL) acts through the long form (LF) of the human PRL receptor (hPRLR) to cause differentiation of mammary epithelial cells through activation of the Janus kinase-2 (JAK2)/signal transducer and activator of transcription 5 (STAT5) pathway and subsequent transcriptional events. To determine whether the inhibitory action of hPRLR short forms (SFs; S1a and S1b) on PRL-induced signal transduction through the LF results from heterodimerization, we studied complex formation among variant forms of the hPRLR. 3'-Tagged fusion constructs, with activities comparable to the wild-type species, were used to investigate homodimer and heterodimer formation. The LF and both SFs of the hPRLR formed homodimers under nonreducing conditions, independently of PRL, but formed only monomers under reducing conditions. Coimmunoprecipitation of the cotransfected LF with the SFs (S1a or S1b) in transfected cells showed ligand-independent heterodimerization of individual SFs with the LF. Bioluminescence resonance energy transfer analysis demonstrated homo- and heterodimeric associations of hPRLR variants in human embryonic kidney 293 cells. Biotin-avidin immunoprecipitation analysis revealed that hPRLR forms are cell surface receptors and that SFs do not influence the steady state or half-life of the LF. Significant homo- and heterodimerization of biotinylated membrane hPRLR forms was observed. These findings indicate that homo- and heterodimers of hPRLR are constitutively present, and that the bivalent hormone acts on the preformed LF homodimer to induce the active signal transduction configuration. Although SF homodimers and their heterodimers with LF mediate JAK2 activation, the SF heterodimer partner lacks cytoplasmic sequences essential for activation of the JAK2/signal transducer and activator of transcription 5 pathway. This prevents the heterodimeric LF from mediating activation of PRL-induced genes.


Subject(s)
Dimerization , Prolactin/pharmacology , Protein Isoforms/metabolism , Receptors, Prolactin/antagonists & inhibitors , Receptors, Prolactin/metabolism , Antigens, Surface/metabolism , Cells, Cultured , Cysteine/metabolism , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins/metabolism , Humans , Janus Kinase 2 , Ligands , Luminescent Proteins/metabolism , Mutagenesis, Site-Directed , Oligopeptides , Peptides/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Recombinant Proteins/metabolism , Transfection
6.
Surgery ; 158(4): 981-6; discussion 986-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26189069

ABSTRACT

PURPOSE: MicroRNA (miR)-26a has been identified as a tumor suppressor in pancreatic cancer cells. Although wild-type p53 controls cell-cycle progression, its mutant form normally present in pancreatic cancer loses this capability. Phosphorylation is known to restore wild-type activity to mutant p53. We, therefore, examined whether miR-26a treatment can restore wild-type functions of mutant p53 via phosphorylation, resulting in inhibition of cell growth. METHODS: The human pancreatic cancer cell line BxPc-3 harboring mutant p53 was used for colony formation, cell-cycle, and Western blotting assays. Gene profile analysis was conducted after transfection with pre-miR-26a. RESULTS: miR-26a expression significantly decreased cell proliferation by 80% along with marked inhibition of colony formation and cell migration. Cell-cycle inhibition at the G0/G1 interface was observed along with enhanced drug retention and increased chemosensitivity to gemcitabine. Mutant p53 was phosphorylated rapidly at its Ser9 and Ser392 residues, but not at Ser15 or Ser20. Gene profile analysis of pre-miR-26a-transfected cells showed a significant increase in gene transcripts promoting apoptosis and p53 activation, with decreased levels of genes involved in cell-cycle progression. CONCLUSION: Delivery of miR-26a may represent a novel strategy for inhibiting pancreatic cancer growth, at least in part by enhancing phosphorylation of mutant p53 to restore its wild-type functions.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation/physiology , MicroRNAs/metabolism , Pancreatic Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Blotting, Western , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle/physiology , Cell Line, Tumor , Cell Movement/physiology , Gene Expression Regulation, Neoplastic , Humans , Mutation , Neoplastic Stem Cells , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation , Transfection , Tumor Suppressor Protein p53/genetics
7.
Pharmaceuticals (Basel) ; 7(1): 46-57, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24451403

ABSTRACT

Recent observations suggest a lower incidence of malignancies in patients infected with HIV during treatment with Highly Active Anti-Retroviral Therapy (HAART) utilizing protease inhibitors. We investigated the effects of ritonavir, a FDA approved HIV protease inhibitor, on proliferation of pancreatic ductal adeno-carcinoma (PDAC) cell lines. Human PDAC cell lines BxPC-3, MIA PaCa-2, and PANC-1 were propagated under standard conditions and treated with serial dilutions of ritonavir. Ritonavir inhibited cell growth in a dose-dependent manner as well as activated the intrinsic apoptotic pathway in human pancreatic ductal adenocarcinoma (PDAC) cell lines. We observed down-modulation of cell-cycle promoting and up-regulation of cell-cycle inhibitory genes; enhanced interaction of retinoblastoma protein (RB) with E2F-1 transcription factor; inhibition of phosphorylation of RB, resulting in sequestration of E2F-1 and subsequent down-regulation of S phase genes; decreased interaction of E2F-1 with its consensus binding sites; inhibition of cell motility and invasiveness; and inhibition of the AKT pathway. Our results demonstrate a potential use of ritonavir as part of combination chemotherapy for PDAC. Since ritonavir is FDA approved for HIV, drug repositioning for PDAC would limit the costs and reduce risks.

8.
CNS Neurol Disord Drug Targets ; 13(3): 533-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24059295

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder that leads to memory problems. It has been associated with type 2 diabetes mellitus at both the molecular and biochemical level. Pancreatic cells have molecular similarities to the brain at the transcriptomic and proteomic levels. Several genes have been reported to be responsible for both AD and diabetes. Currently, no proper treatment is available but various therapeutic approaches are utilized worldwide for the management of these disorders and may be nanoparticles and herbal treatment of Bacopa monnieri will make promise for the treatment of AD in future. The formation of amyloids in neurons and the formation of amylin in pancreatic cells are potential links between these two disorders, which can be silent killers.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Gene Expression Profiling , Humans , Proteomics
9.
Surgery ; 154(4): 739-46; discussion 746-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24074410

ABSTRACT

PURPOSE: Enhancer of zeste homologue 2 (EZH2), a component of the chromatin modification protein complex, is upregulated in pancreatic ductal adenocarcinoma (PDAC), whereas loss of p53 and its downstream target, p21(waf1/cip1), is also observed frequently. We sought to investigate the role of the p53-p21(waf1/cip1) pathway in relation to EZH2-mediated inhibition of PDAC. METHODS: The PANC-1 cell line was utilized in chromatin immunoprecipitation, gene profiling, Western blot, cell invasion, cell proliferation, and tumor xenograft assays. RESULTS: Western blot analysis with antibodies that recognize both wild-type and mutant p53 did not show any alterations in band intensity; however, antibody that detects only mutant p53 showed a band of significantly lesser intensity with EZH2 knockdown. Western blot analysis further revealed a significant upregulation of p21(waf1/cip1). Gene expression profile analysis indicated significantly enhanced transcripts of transcriptional inducers of p21(waf1/cip1), with downregulation of mutant p53 transcript, corroborating the Western blot analysis. PANC-1 cells expressing EZH2-short hairpin RNA displayed markedly attenuated growth in SCID mice. CONCLUSION: Downregulation of mutant p53 with concomitant enhanced expression of p21(waf1/cip1) and its transcriptional trans-activators may contribute toward EZH2-mediated suppression of PDAC.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Genes, p53/physiology , Pancreatic Neoplasms/genetics , Polycomb Repressive Complex 2/physiology , RNA, Small Interfering/genetics , Adenocarcinoma/pathology , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enhancer Elements, Genetic , Enhancer of Zeste Homolog 2 Protein , Humans , Mice , Pancreatic Neoplasms/pathology , Polycomb Repressive Complex 2/genetics , Up-Regulation
10.
Cancer Lett ; 336(1): 53-60, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23603558

ABSTRACT

The enhancer of zeste homolog 2 (EZH2) methyltransferase is a transcriptional repressor. EZH2 is abnormally elevated in epithelial ovarian cancer (EOC). We demonstrated that EZH2 knockdown inhibited cell growth, activated apoptosis, and enhanced chemosensitivity. Further, silencing of EZH2 resulted in re-expression of p21(waf1/cip1) and down-regulation of mutant p53. Finally, EZH2 knockdown contributed to attenuated EOC growth in SCID mice.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/pathology , Polycomb Repressive Complex 2/metabolism , RNA Interference , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enhancer of Zeste Homolog 2 Protein , Female , Humans , Mice , Mice, SCID , Mutation , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , Polycomb Repressive Complex 2/genetics , Tumor Suppressor Protein p53/genetics
11.
Surgery ; 152(4): 704-11; discussion 711-3, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22943841

ABSTRACT

PURPOSE: To investigate the possibility of inhibiting the progression of pancreatic ductal adenocarcinoma (PDAC) by facilitating the expression of E-cadherin through the enforced expression of microRNA-101 (miR-101). METHODS: In situ hybridization was conducted with archival tissue using a double digoxigenin-labeled probe. Chromatin immunoprecipitation (ChIP) assay was conducted with EZ-Magna ChIPTM A. Gene profile analysis, Western blot, and immunoprecipitation assays were performed using standard protocols. RESULTS: We found that decreased miR-101 expression observed in archival patient tissues was significantly associated with poor prognosis indicated by low-intensity staining in high-grade tumors. ChIP assays using anti-enhancer of zeste homolog 2 (EZH2) antibodies indicated not only the interaction of EZH2 to the CDH1 (E-cadherin) promoter, but also that this interaction was significantly diminished in cells transfected with pre-miR-101. We observed a global downregulation of trimethylated lysine 27 of H3 histone (H3K27me3) along with upregulation of the enzymes histone deacetylase -1 and -2 with the re-expression of miR-101. Further, we observed lesser levels of transcriptional factors that inhibit the CDH1 promoter with pre-miR-101 treatment. Western blot analysis confirmed the enhanced E-cadherin expression. PANC-1 cells transduced with pre-miR-101 displayed markedly attenuated growth in SCID mice. CONCLUSION: These results suggest the potential therapeutic use of miR-101-enforced expression for inhibition of PDAC.


Subject(s)
Cadherins/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/therapy , MicroRNAs/therapeutic use , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Adult , Aged , Animals , Antigens, CD , Apoptosis/genetics , Base Sequence , Carcinoma, Pancreatic Ductal/pathology , Case-Control Studies , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Female , Gene Expression , Humans , In Situ Hybridization , Male , Mice , Mice, SCID , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness/genetics , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Transduction, Genetic , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
13.
Methods Mol Biol ; 755: 245-56, 2011.
Article in English | MEDLINE | ID: mdl-21761309

ABSTRACT

Pure populations of tumor cells are essential for the identification of tumor-associated proteins for the development of targeted therapy. In recent years, laser capture microdissection (LCM) has been used successfully to obtain distinct populations of cells for subsequent molecular analysis. The polycomb group (PcG) protein, enhancer of zeste homolog 2 (EzH2), a methyl-transferase that plays a key role in -transcriptional gene repression, is frequently overexpressed in several malignant tumors. High levels of EzH2 are often associated with advanced disease stage in many solid tumors; however, its role in the pathogenesis of pancreatic ductal adeno-carcinoma (PDAC) is poorly understood. Because of the limited sample availability and the absence of in vitro amplification steps for proteins, the use of LCM for proteomics studies largely depends on highly sensitive protein detection methods. Here, we developed a faster and sensitive Western blot protocol and validated it for the detection of EzH2 in ∼2,000 cells. Initially, cultured PANC-1 cells were used to optimize protein electrophoresis and western blotting conditions. Gradient gel electrophoresis in combination with optimized antibody concentrations, and a sensitive chemiluminescent assay provided a strong signal. In order to further confirm the role of EzH2 in PDAC, employing siRNA-mediated gene silencing via long lasting plasmid vectors containing shRNA, we investigated the potential role of EzH2 gene silencing in pancreatic cancer regression. Positive correlation of EzH2 expression was observed with advanced stage, serous histology, and increasing grade in pancreatic cancer patient tissues. Further EzH2 knockdown resulted in decreased cell growth and invasiveness. The findings of this study emphasize that western blotting of a LCM-generated pure population of cancer cells may be a valuable technique for the study of tumor-specific proteins.


Subject(s)
Blotting, Western/methods , Carcinoma, Pancreatic Ductal/metabolism , DNA-Binding Proteins/metabolism , Lasers , Microdissection/methods , Pancreatic Neoplasms/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Migration Assays/methods , Cell Proliferation , Cell Separation/methods , Cryopreservation , Electrophoresis, Polyacrylamide Gel/methods , Enhancer of Zeste Homolog 2 Protein , Humans , Microtomy/methods , Pancreatic Neoplasms/pathology , Polycomb Repressive Complex 2 , Rats , Staining and Labeling/methods , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL